Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters








Publication year range
1.
BMC Res Notes ; 16(1): 54, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37069662

ABSTRACT

OBJECTIVE: A Delta-Notch signaling component, Notch1, is involved in the normal development and multiple disorders of the kidney. Although the increase in Notch1 signaling is crucial to these pathogeneses, the basal signaling level in 'healthy' mature kidneys is still unclear. To address this question, we used an artificial Notch1 receptor fused with Gal4/UAS components in addition to the Cre/loxP system and fluorescent proteins in mice. This transgenic reporter mouse system enabled labeling of past and ongoing Notch1 signaling with tdsRed or Cre recombinase, respectively. RESULTS: We confirmed that our transgenic reporter mouse system mimicked the previously reported Notch1 signaling pattern. Using this successful system, we infrequently observed cells with ongoing Notch1 signaling only in Bowman's capsule and tubules. We consider that Notch1 activation in several lines of disease model mice was pathologically significant itself.


Subject(s)
Health , Kidney , Receptor, Notch1 , Signal Transduction , Animals , Mice , Kidney/cytology , Kidney/metabolism , Ligands , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Epithelial Cells/metabolism , Bowman Capsule/cytology , Bowman Capsule/metabolism , Attachment Sites, Microbiological , Genes, Reporter , Receptor, Notch1/genetics , Receptor, Notch1/metabolism
2.
JCI Insight ; 6(24)2021 12 22.
Article in English | MEDLINE | ID: mdl-34767537

ABSTRACT

Kidneys are critical target organs of COVID-19, but susceptibility and responses to infection remain poorly understood. Here, we combine SARS-CoV-2 variants with genome-edited kidney organoids and clinical data to investigate tropism, mechanism, and therapeutics. SARS-CoV-2 specifically infects organoid proximal tubules among diverse cell types. Infections produce replicating virus, apoptosis, and disrupted cell morphology, features of which are revealed in the context of polycystic kidney disease. Cross-validation of gene expression patterns in organoids reflects proteomic signatures of COVID-19 in the urine of critically ill patients indicating interferon pathway upregulation. SARS-CoV-2 viral variants alpha, beta, gamma, kappa, and delta exhibit comparable levels of infection in organoids. Infection is ameliorated in ACE2-/- organoids and blocked via treatment with de novo-designed spike binder peptides. Collectively, these studies clarify the impact of kidney infection in COVID-19 as reflected in organoids and clinical populations, enabling assessment of viral fitness and emerging therapies.


Subject(s)
Acute Kidney Injury/urine , COVID-19/urine , Kidney Tubules, Proximal/virology , Kidney/virology , Organoids/virology , SARS-CoV-2/pathogenicity , Acute Kidney Injury/etiology , Adult , Aged , Angiotensin-Converting Enzyme 2/genetics , Animals , Apoptosis , Bowman Capsule/cytology , Bowman Capsule/virology , COVID-19/complications , Chlorocebus aethiops , Female , Gene Knockout Techniques , Hospital Mortality , Hospitalization , Humans , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Middle Aged , Organoids/metabolism , Podocytes/virology , Polycystic Kidney Diseases , Protein Kinase D2/genetics , Proteome , Receptors, Coronavirus/genetics , Reproducibility of Results , Transcriptome , Vero Cells , Viral Tropism , Virus Replication
3.
Cell Tissue Res ; 380(3): 581-591, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31989254

ABSTRACT

Phenotypic changes in culture hamper the identification and characterization of cultured podocytes and parietal epithelial cells of the Bowman's capsule (PECs). We have recently established culture conditions that restore podocytes to their differentiated phenotypes. We compared podocytes and PECs cultured under the same conditions to determine the unique characteristics of the two cell types. Performing this comparison under the same conditions accentuated these differences. Podocytes behaved like non-epithelial cells by extending cell processes even at confluence. By contrast, PECs behaved like typical epithelial cells by maintaining a polygonal appearance. Other differences were identified using immunostaining and RT-PCR; podocytes expressed high levels of podocyte-specific markers while PECs expressed high levels of PEC-specific markers. However, while podocytes expressed low levels of PEC markers, PECs expressed low levels of podocyte markers. Therefore, the identification of podocytes and PECs in culture requires the evaluation of respective cell markers and the expression of markers for other cell types.


Subject(s)
Bowman Capsule/cytology , Epithelial Cells/cytology , Podocytes/cytology , Animals , Biomarkers/metabolism , Cells, Cultured , Rats
4.
Kidney Int ; 96(1): 80-93, 2019 07.
Article in English | MEDLINE | ID: mdl-31029503

ABSTRACT

Beside the classical flat parietal epithelial cells (PECs), we investigated proximal tubular epithelial-like cells, a neglected subgroup of PECs. These cells, termed cuboidal PECs, make up the most proximal part of the proximal tubule and may also line parts of Bowman's capsule. Additionally, a third intermediate PEC subgroup was identified at the junction between the flat and cuboidal PEC subgroups at the tubular orifice. The transgenic mouse line PEC-rtTA labeled all three PEC subgroups. Here we show that the inducible Pax8-rtTA mouse line specifically labeled only cuboidal and intermediate PECs, but not flat PECs. In aging Pax8-rtTA mice, cell fate mapping showed no evidence for significant transdifferentiation from flat PECs to cuboidal or intermediate PECs or vice versa. In murine glomerular disease models of crescentic glomerulonephritis, and focal segmental glomerulosclerosis (FSGS), intermediate PECs became more numerous. These intermediate PECs preferentially expressed activation markers CD44 and Ki-67, suggesting that this subgroup of PECs was activated more easily than the classical flat PECs. In mice with FSGS, cuboidal and intermediate PECs formed sclerotic lesions. In patients with FSGS, cells forming the tip lesions expressed markers of intermediate PECs. These novel PEC subgroups form sclerotic lesions and were more prone to cellular activation compared to the classical flat PECs in disease. Thus, colonization of Bowman's capsule by cuboidal PECs may predispose to lesion formation and chronic kidney disease. We propose that tip lesions originate from this novel subgroup of PECs in patients with FSGS.


Subject(s)
Epithelial Cells/pathology , Glomerulosclerosis, Focal Segmental/pathology , Kidney Glomerulus/pathology , Kidney Tubules, Proximal/cytology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Bowman Capsule/cytology , Disease Models, Animal , Epithelial Cells/metabolism , Female , Humans , Hyaluronan Receptors/metabolism , Ki-67 Antigen/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , PAX8 Transcription Factor/genetics , Young Adult
5.
Kidney Int ; 92(6): 1444-1457, 2017 12.
Article in English | MEDLINE | ID: mdl-28756872

ABSTRACT

Proliferation of glomerular epithelial cells, including podocytes, is a key histologic feature of crescentic glomerulonephritis. We previously found that retinoic acid (RA) inhibits proliferation and induces differentiation of podocytes by activating RA receptor-α (RARα) in a murine model of HIV-associated nephropathy. Here, we examined whether RA would similarly protect podocytes against nephrotoxic serum-induced crescentic glomerulonephritis and whether this effect was mediated by podocyte RARα. RA treatment markedly improved renal function and reduced the number of crescentic lesions in nephritic wild-type mice, while this protection was largely lost in mice with podocyte-specific ablation of Rara (Pod-Rara knockout). At a cellular level, RA significantly restored the expression of podocyte differentiation markers in nephritic wild-type mice, but not in nephritic Pod-Rara knockout mice. Furthermore, RA suppressed the expression of cell injury, proliferation, and parietal epithelial cell markers in nephritic wild-type mice, all of which were significantly dampened in nephritic Pod-Rara knockout mice. Interestingly, RA treatment led to the coexpression of podocyte and parietal epithelial cell markers in a small subset of glomerular cells in nephritic mice, suggesting that RA may induce transdifferentiation of parietal epithelial cells toward a podocyte phenotype. In vitro, RA directly inhibited the proliferation of parietal epithelial cells and enhanced the expression of podocyte markers. In vivo lineage tracing of labeled parietal epithelial cells confirmed that RA increased the number of parietal epithelial cells expressing podocyte markers in nephritic glomeruli. Thus, RA attenuates crescentic glomerulonephritis primarily through RARα-mediated protection of podocytes and in part through the inhibition of parietal epithelial cell proliferation and induction of their transdifferentiation into podocytes.


Subject(s)
Cell Proliferation/drug effects , Glomerulonephritis/drug therapy , Podocytes/drug effects , Protective Agents/pharmacology , Retinoic Acid Receptor alpha/metabolism , Tretinoin/pharmacology , Animals , Autoantibodies/administration & dosage , Autoantibodies/immunology , Biomarkers/metabolism , Biopsy , Bowman Capsule/cytology , Bowman Capsule/drug effects , Bowman Capsule/physiology , Cell Transdifferentiation/drug effects , Cells, Cultured , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Podocytes/pathology , Podocytes/physiology , Protective Agents/therapeutic use , Retinoic Acid Receptor alpha/genetics , Tretinoin/therapeutic use
6.
Adv Anat Pathol ; 24(1): 24-34, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27677082

ABSTRACT

The glomerulus has 3 resident cells namely mesangial cells that produce the mesangial matrix, endothelial cells that line the glomerular capillaries, and podocytes that cover the outer surface of the glomerular basement membrane. Parietal epithelial cells (PrECs), which line the Bowman's capsule are not part of the glomerular tuft but may have an important role in the normal function of the glomerulus. A significant progress has been made in recent years regarding our understanding of the role and function of these cells in normal kidney and in kidneys with various types of glomerulopathy. In crescentic glomerulonephritis necrotizing injury of the glomerular tuft results in activation and leakage of fibrinogen which provides the trigger for excessive proliferation of PrECs giving rise to glomerular crescents. In cases of collapsing glomerulopathy, podocyte injury causes collapse of the glomerular capillaries and activation and proliferation of PrECs, which accumulate within the urinary space in the form of pseudocrescents. Many of the noninflammatory glomerular lesions such as focal segmental glomerulosclerosis and global glomerulosclerosis also result from podocyte injury which causes variable loss of podocytes. In these cases podocyte injury leads to activation of PrECs that extend on to the glomerular tuft where they cause segmental and/or global sclerosis by producing excess matrix, resulting in obliteration of the capillary lumina. In diabetic nephropathy, in addition to increased matrix production in the mesangium and glomerular basement membranes, increased loss of podocytes is an important determinant of long-term prognosis. Contrary to prior belief there is no convincing evidence for an active podocyte proliferation in any of the above mentioned glomerulopathies.


Subject(s)
Cell Communication/physiology , Epithelial Cells , Kidney , Podocytes , Animals , Bowman Capsule/cytology , Humans
7.
Kidney Int ; 89(6): 1221-30, 2016 06.
Article in English | MEDLINE | ID: mdl-27165817

ABSTRACT

Podocytes maintain the glomerular filtration barrier, and the stability of this barrier depends on their highly differentiated postmitotic phenotype, which also defines the particular vulnerability of the glomerulus. Recent podocyte biology and gene disruption studies in vivo indicate a causal relationship between abnormalities of single podocyte molecules and proteinuria and glomerulosclerosis. Podocytes live under various stresses and pathological stimuli. They adapt to maintain homeostasis, but excessive stress leads to maladaptation with complex biological changes including loss of integrity and dysregulation of cellular metabolism. Podocyte injury causes proteinuria and detachment from the glomerular basement membrane. In addition to "sick" podocytes and their detachment, our understanding of glomerular responses following podocyte loss needs to address the pathways from podocyte injury to sclerosis. Studies have found a variety of glomerular responses to podocyte dysfunction in vivo, such as disruption of podocyte-endothelial cross talk and activation of podocyte-parietal cell interactions, all of which help us to understand the complex scenario of podocyte injury and its consequences. This review focuses on the cellular aspects of podocyte dysfunction and the adaptive or maladaptive glomerular responses to podocyte injury that lead to its major consequence, glomerulosclerosis.


Subject(s)
Glomerular Basement Membrane/pathology , Glomerulosclerosis, Focal Segmental/pathology , Podocytes/pathology , Proteinuria/pathology , Apoptosis , Bowman Capsule/cytology , Bowman Capsule/pathology , Cell Communication , Cell Differentiation , Endothelial Cells , Glomerular Basement Membrane/cytology , Glomerular Basement Membrane/metabolism , Humans , Oxidative Stress , Podocytes/cytology , Podocytes/metabolism , Sclerosis , Stress, Mechanical
8.
J Am Soc Nephrol ; 26(6): 1389-401, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25270065

ABSTRACT

Parietal epithelial cells have been identified as potential progenitor cells in glomerular regeneration, but the molecular mechanisms underlying this process are not fully defined. Here, we established an immortalized polyclonal human parietal epithelial cell (hPEC) line from naive human Bowman's capsule cells isolated by mechanical microdissection. These hPECs expressed high levels of PEC-specific proteins and microRNA-193a (miR-193a), a suppressor of podocyte differentiation through downregulation of Wilms' tumor 1 in mice. We then investigated the function of miR-193a in the establishment of podocyte and PEC identity and determined whether inhibition of miR-193a influences the behavior of PECs in glomerular disease. After stable knockdown of miR-193a, hPECs adopted a podocyte-like morphology and marker expression, with decreased expression levels of PEC markers. In mice, inhibition of miR-193a by complementary locked nucleic acids resulted in an upregulation of the podocyte proteins synaptopodin and Wilms' tumor 1. Conversely, overexpression of miR-193a in vivo resulted in the upregulation of PEC markers and the loss of podocyte markers in isolated glomeruli. Inhibition of miR-193a in a mouse model of nephrotoxic nephritis resulted in reduced crescent formation and decreased proteinuria. Together, these results show the establishment of a human PEC line and suggest that miR-193a functions as a master switch, such that glomerular epithelial cells with high levels of miR-193a adopt a PEC phenotype and cells with low levels of miR-193a adopt a podocyte phenotype. miR-193a-mediated maintenance of PECs in an undifferentiated reactive state might be a prerequisite for PEC proliferation and migration in crescent formation.


Subject(s)
Cell Transdifferentiation/genetics , Gene Expression Regulation , Glomerulosclerosis, Focal Segmental/genetics , MicroRNAs/genetics , Podocytes/metabolism , Animals , Bowman Capsule/cytology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , Fluorescent Antibody Technique , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/physiopathology , Mice , Mice, Transgenic , Phenotype , Polymerase Chain Reaction/methods , Random Allocation , Statistics, Nonparametric
9.
J Membr Biol ; 247(8): 675-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24906870

ABSTRACT

In kidney nephron, parietal epithelial cells line the Bowman's capsule and function as a permeability barrier for the glomerular filtrate. Bowman's capsule cells with proximal tubule epithelial morphology have been found. However, the effects of tubular metaplasia in Bowman's capsule on kidney function remain poorly understood. Sodium-glucose cotransporter 2 (SGLT2) plays a major role in reabsorption of glucose in the kidney and is expressed on brush border membrane (BBM) of epithelial cells in the early segment of the proximal tubule. We hypothesized that SGLT2 is expressed in tubularized Bowman's capsule and used our novel antibody to test this hypothesis. Immunohistochemical analysis was performed with our SGLT2 antibody on C57BL/6 mouse kidney prone to have tubularized Bowman's capsules. Cell membrane was examined with periodic acid-Schiff (PAS) stain. The results showed that SGLT2 was localized on BBM of the proximal tubules in young and adult mice. Bowman's capsules were lined mostly with normal brush border-less parietal epithelial cells in young mice, while they were almost completely covered with proximal tubule-like cells in adult mice. Regardless of age, SGLT2 was expressed on BBM of the tubularized Bowman's capsule but did not co-localize with nephrin in the glomerulus. SGLT2-expressing tubular cells expanded from the urinary pole toward the vascular pole of the Bowman's capsule. This study identified the localization of SGLT2 in the Bowman's capsule. Bowman's capsules with tubular metaplasia may acquire roles in reabsorption of filtered glucose and sodium.


Subject(s)
Bowman Capsule/metabolism , Cell Membrane/metabolism , Epithelium/metabolism , Kidney Tubules, Proximal/metabolism , Kidney/metabolism , Microvilli/metabolism , Sodium-Glucose Transporter 2/metabolism , Animals , Blotting, Western , Bowman Capsule/cytology , Glucose/metabolism , Immunoenzyme Techniques , Kidney/cytology , Kidney Tubules, Proximal/cytology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Sodium/metabolism
10.
Nephron Exp Nephrol ; 126(2): 91, 2014.
Article in English | MEDLINE | ID: mdl-24854648

ABSTRACT

BACKGROUND: The incidence of kidney diseases is increasing worldwide and they are emerging as a major public health problem. Once mostly considered inexorable, renal disease progression can now be halted and lesions can even regress with drugs such as angiotensin-converting enzyme inhibitors (ACEi) and angiotensin II type I receptor blockers, indicating the possibility of kidney repair. SUMMARY: The discovery of renal progenitor cells lining the Bowman capsule of adult rat and human kidneys has shed light on the mechanism of repair by ACEi. Parietal progenitors are a reservoir of cells that contribute to podocyte turnover in physiological conditions. In the early phases of renal disease these progenitors migrate chaotically and subsequently proliferate, accumulating in Bowman's space. The abnormal behavior of parietal progenitors is sustained by the activation of CXCR4 receptors in response to an increased production of the chemokine SDF-1 by podocytes activated by the inflammatory environment. Ang II, via the AT1 receptor, also contributes to progenitor cell proliferation. The CXCR4/SDF-1 and Ang II/AT1 receptor pathogenic pathways both pave the way for lesion formation and subsequent sclerosis. ACEi normalize the CXCR4 and AT1 receptor expression on progenitors, limiting their proliferation, concomitant with the regression of hyperplastic lesions in animals, and in a patient with crescentic glomerulopathy. KEY MESSAGE: Understanding the molecular and cellular determinants of regeneration triggered by renoprotective drugs will reveal novel pathways that might be challenged or targeted by pharmacological therapy.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Kidney/physiology , Regeneration/drug effects , Angiotensin II/physiology , Animals , Bowman Capsule/cytology , Chemokine CXCL12 , Humans , Podocytes/physiology , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/physiology , Receptors, CXCR4/drug effects , Receptors, CXCR4/physiology , Regeneration/physiology , Stem Cells/physiology
11.
Methods Cell Biol ; 121: 121-39, 2014.
Article in English | MEDLINE | ID: mdl-24560507

ABSTRACT

Cells live in a highly curved and folded micropatterned environment within the human body. Hence, there is a need to develop engineering paradigms to replicate these microenvironments in order to investigate the behavior of cells in vitro, as well as to develop bioartificial organs for tissue engineering and regenerative medicine. In this chapter, we first motivate the need for such micropatterns based on anatomical considerations and then survey methods that can be utilized to generate curved and folded micropatterns of relevance to 3D cell culture and tissue engineering. The methods surveyed can broadly be divided into two classes: top-down approaches inspired by conventional 2D microfabrication and bottom-up approaches most notably in the self-assembly of thin patterned films. These methods provide proof of concept that the high resolution, precise and reproducible patterning of cell and matrix microenvironments in anatomically relevant curved and folded geometries is possible. A specific protocol is presented to create curved and folded hydrogel micropatterns.


Subject(s)
Cellular Microenvironment/physiology , Coated Materials, Biocompatible , Tissue Engineering/methods , Animals , Biocompatible Materials , Bowman Capsule/blood supply , Bowman Capsule/cytology , Bowman Capsule/physiology , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Ear/physiology , Humans , Intestine, Small/cytology , Intestine, Small/physiology , Kidney Glomerulus/blood supply , Kidney Glomerulus/cytology , Kidney Glomerulus/physiology , Microtechnology , Podocytes/physiology , Rats , Surface Properties
12.
Hum Pathol ; 45(2): 382-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24439225

ABSTRACT

Acute tubular necrosis (ATN), elicited by ischemia and/or toxicity, is a potentially life-threatening condition. Histologically, ATN corresponds to necrosis and detachment of renal tubular epithelial cells. However, the tubules possess a considerable regenerative capacity and may be restored. We have previously identified a scattered population of progenitor-like cells within the proximal tubules, sharing marker expression with the parietal epithelial cells of Bowman's capsule as well as with renal tubules regenerating after ATN. In the present analysis, we use transmission electron microscopy, immunoelectron microscopy and immunofluorescence of human kidney cortex to further explore these cells. We demonstrate that the cells are smaller and have drastically fewer mitochondria than the surrounding proximal tubule cells. They also display strong expression of several structural proteins such as vimentin, collagen-7A1 and the tight junction protein claudin-1. To functionally assess these cells, we also developed a novel human kidney explant model of ATN demonstrating that the cells are more resilient to injury than the surrounding proximal tubular cells. Taken together the results suggest a novel robust cell type with a contrasting biological role to that of the bulk of proximal tubular epithelium.


Subject(s)
Kidney Tubules, Proximal/cytology , Stem Cells/metabolism , Bowman Capsule/cytology , Claudin-1/biosynthesis , Collagen Type VII/biosynthesis , Epithelium/metabolism , Humans , Kidney/cytology , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubules, Proximal/ultrastructure , Vimentin/biosynthesis
13.
J Am Soc Nephrol ; 25(4): 693-705, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24408873

ABSTRACT

Previously, we showed that some podocytes in juvenile mice are recruited from cells lining Bowman's capsule, suggesting that parietal epithelial cells (PECs) are a progenitor cell population for podocytes. To investigate whether PECs also replenish podocytes in adult mice, PECs were genetically labeled in an irreversible fashion in 5-week-old mice. No significant increase in labeled podocytes was observed, even after 18 months. To accelerate a potential regenerative mechanism, progressive glomerular hypertrophy was induced by progressive partial nephrectomies. Again, no significant podocyte replenishment was observed. Rather, labeled PECs exclusively invaded segments of the tuft affected by glomerulosclerosis, consistent with our previous findings. We next reassessed PEC recruitment in juvenile mice using a different reporter mouse and confirmed significant recruitment of labeled PECs onto the glomerular tuft. Moreover, some labeled cells on Bowman's capsule expressed podocyte markers, and cells on Bowman's capsule were also directly labeled in juvenile podocyte-specific Pod-rtTA transgenic mice. In 6-week-old mice, however, cells on Bowman's capsule no longer expressed podocyte-specific markers. Similarly, in human kidneys, some cells on Bowman's capsule expressed the podocyte marker synaptopodin from 2 weeks to 2 years of age but not at 7 years of age. In summary, podocyte regeneration from PECs could not be detected in aging mice or models of glomerular hypertrophy. We propose that a small fraction of committed podocytes reside on Bowman's capsule close to the vascular stalk and are recruited onto the glomerular tuft during infancy to adolescence in mice and humans.


Subject(s)
Epithelial Cells/physiology , Podocytes/cytology , Regeneration/physiology , Aging/pathology , Animals , Bowman Capsule/cytology , Glomerulosclerosis, Focal Segmental/pathology , Hypertrophy , Kidney Glomerulus/pathology , Mice
14.
Histochem Cell Biol ; 141(5): 459-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24429831

ABSTRACT

A special feature of the renal stem/progenitor cell niche is its always close neighborhood to the capsule during organ development. To explore this link, neonatal kidney was investigated by histochemistry and transmission electron microscopy. For adequate contrasting, fixation of specimens was performed by glutaraldehyde including tannic acid. The immunohistochemical data illustrate that renal stem/progenitor cells are not distributed randomly but are positioned specially to the capsule. Epithelial stem/progenitor cells are found to be enclosed by the basal lamina at a collecting duct (CD) ampulla tip. Only few layers of mesenchymal cells are detected between epithelial cells and the capsule. Most impressive, numerous microfibers reacting with soybean agglutinin, anti-collagen I and III originate from the basal lamina at a CD ampulla tip and line between mesenchymal stem/progenitor cells to the inner side of the capsule. This specific arrangement holds together both types of stem/progenitor cells in a cage and fastens the niche as a whole at the capsule. Electron microscopy further illustrates that the stem/progenitor cell niche is in contact with a tunnel system widely spreading between atypical smooth muscle cells at the inner side of the capsule. It seems probable that stem/progenitor cells are supplied here by interstitial fluid.


Subject(s)
Bowman Capsule/cytology , Bowman Capsule/ultrastructure , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/ultrastructure , Stem Cell Niche , Animals , Female , Male , Microscopy, Electron, Transmission , Rabbits
15.
J Am Soc Nephrol ; 25(1): 129-41, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24071005

ABSTRACT

Parietal podocytes are fully differentiated podocytes lining Bowman's capsule where normally only parietal epithelial cells (PECs) are found. Parietal podocytes form throughout life and are regularly observed in human biopsies, particularly in atubular glomeruli of diseased kidneys; however, the origin of parietal podocytes is unresolved. To assess the capacity of PECs to transdifferentiate into parietal podocytes, we developed and characterized a novel method for creating atubular glomeruli by electrocoagulation of the renal cortex in mice. Electrocoagulation produced multiple atubular glomeruli containing PECs as well as parietal podocytes that projected from the vascular pole and lined Bowman's capsule. Notably, induction of cell death was evident in some PECs. In contrast, Bowman's capsules of control animals and normal glomeruli of electrocoagulated kidneys rarely contained podocytes. PECs and podocytes were traced by inducible and irreversible genetic tagging using triple transgenic mice (PEC- or Pod-rtTA/LC1/R26R). Examination of serial cryosections indicated that visceral podocytes migrated onto Bowman's capsule via the vascular stalk; direct transdifferentiation from PECs to podocytes was not observed. Similar results were obtained in a unilateral ureter obstruction model and in human diseased kidney biopsies, in which overlap of PEC- or podocyte-specific antibody staining indicative of gradual differentiation did not occur. These results suggest that induction of atubular glomeruli leads to ablation of PECs and subsequent migration of visceral podocytes onto Bowman's capsule, rather than transdifferentiation from PECs to parietal podocytes.


Subject(s)
Kidney Glomerulus/cytology , Podocytes/cytology , Animals , Bowman Capsule/cytology , Cell Lineage , Cell Movement , Cell Transdifferentiation , Disease Models, Animal , Electrocoagulation , Epithelial Cells/cytology , Female , Humans , Kidney Glomerulus/surgery , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Podocytes/metabolism , Ureteral Obstruction/pathology
16.
Am J Physiol Renal Physiol ; 304(4): F333-47, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23235479

ABSTRACT

Progressive loss of podocytes is the most frequent cause accounting for end-stage renal failure. Podocytes are complex, terminally differentiated cells incapable of replicating. Thus lost podocytes cannot be replaced by proliferation of neighboring undamaged cells. Moreover, podocytes occupy a unique position as epithelial cells, adhering to the glomerular basement membrane (GBM) only by their processes, whereas their cell bodies float within the filtrate in Bowman's space. This exposes podocytes to the danger of being lost by detachment as viable cells from the GBM. Indeed, podocytes are continually excreted as viable cells in the urine, and the rate of excretion dramatically increases in glomerular diseases. Given this situation, it is likely that evolution has developed particular mechanisms whereby podocytes resist cell detachment. Podocytes respond to stress and injury by undergoing tremendous changes in shape. Foot process effacement is the most prominent and, yet in some ways, the most enigmatic of those changes. This review summarizes the various structural responses of podocytes to injury, focusing on foot process effacement and detachment. We raise the hypothesis that foot process effacement represents a protective response of podocytes to escape detachment from the GBM.


Subject(s)
Podocytes/physiology , Stress, Physiological/physiology , Animals , Bowman Capsule/cytology , Bowman Capsule/physiopathology , Glomerular Basement Membrane/cytology , Glomerular Basement Membrane/physiology , Humans , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Mice , Podocytes/cytology , Rats
18.
Contrib Nephrol ; 169: 23-36, 2011.
Article in English | MEDLINE | ID: mdl-21252509

ABSTRACT

Parietal epithelial cells of Bowman's capsules were first described by Sir William Bowman in 1842 in his paper On the Structure and Use of the Malpighian Bodies of the Kidney [London, Taylor, 1842], but since then their functions have remained poorly understood. A large body of evidence has recently suggested that parietal epithelial cells represent a reservoir of renal progenitors in adult human kidney which generate novel podocytes during childhood and adolescence, and can regenerate injured podocytes. The discovery that parietal epithelial cells represent a potential source for podocyte regeneration suggests that podocyte injury can be repaired. However, recent results also suggest that an abnormal proliferative response of renal progenitors to podocyte injury can generate hyperplastic glomerular lesions that are observed in crescentic glomerulonephritis and other types of glomerular disorders. Taken together, these results establish an entirely novel view that changes the way of thinking about renal physiology and pathophysiology, and suggest that understanding how self-renewal and fate decision of parietal epithelial cells in response to podocyte injury may be perturbed or modulated will be crucial for obtaining novel tools for prevention and treatment of glomerulosclerosis.


Subject(s)
Bowman Capsule/physiology , Epithelial Cells/physiology , Kidney Diseases/physiopathology , Bowman Capsule/cytology , Cell Communication/physiology , Epithelial Cells/cytology , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Humans , Kidney Diseases/pathology , Podocytes/cytology , Podocytes/physiology
19.
Physiol Genomics ; 43(3): 161-73, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21081658

ABSTRACT

To gain molecular insight into kidney function, we performed a high-resolution quantitative analysis of gene expression in glomeruli and nine different nephron segments dissected from mouse kidney using Serial Analysis of Gene Expression (SAGE). We also developed dedicated bioinformatics tools and databases to annotate mRNA tags as transcripts. Over 800,000 mRNA SAGE tags were sequenced corresponding to >20,000 different mRNA tags present at least twice in at least one library. Hierarchical clustering analysis of tags demonstrated similarities between the three anatomical subsegments of the proximal tubule, between the cortical and medullary segments of the thick ascending limb of Henle's loop, and between the three segments constituting the aldosterone-sensitive distal nephron segments, whereas the glomerulus and distal convoluted tubule clusterized independently. We also identified highly specific mRNA markers of each subgroup of nephron segments and of most nephron segments. Tag annotation also identified numbers of putative antisense mRNAs. This database constitutes a reference resource in which the quantitative expression of a given gene can be compared with that of other genes in the same nephron segment, or between different segments of the nephron. To illustrate possible applications of this database, we performed a deeper analysis of the glomerulus transcriptome that unexpectedly revealed expression of several ion and water carriers; within the glomerulus, they were found to be preferentially expressed in the parietal sheet. It also revealed the major role of the zinc finger transcription factor Wt1 in the specificity of gene expression in the glomerulus. Finally, functional annotation of glomerulus-specific transcripts suggested a high proliferation activity of glomerular cells. Immunolabeling for PCNA confirmed a high percentage of proliferating cells in the glomerulus parietal sheet.


Subject(s)
Gene Expression Profiling , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Animals , Biomarkers/metabolism , Bowman Capsule/cytology , Bowman Capsule/metabolism , Cell Proliferation , Cluster Analysis , Databases, Genetic , Gene Expression Regulation , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Organ Specificity/genetics , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
20.
Int J Biochem Cell Biol ; 42(9): 1380-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20542138

ABSTRACT

In the past decade, podocyte research has been greatly aided by the development of powerful new molecular, cellular and animal tools, leading to elucidation of an increasing number of proteins involved in podocyte function and identification of mutated genes in hereditary glomerulopathies. Accumulating evidence indicates that podocyte disorders may not only underlie these hereditary glomerulopathies but also play crucial role in a broad spectrum of acquired glomerular diseases. Genetic susceptibility, environmental influence and systemic responses are all involved in the mediation of the pathogenesis of podocytopathies. Injured podocytes may predisopose to further injury of other podocytes and other adjacent/distant renal cells in a vicious cycle, leading to inexorable progression of glomerular injury. The classic view is that podocytes have a limited ability to proliferate in the normal mature kidney. However, recent research in rodents has provided suggestive evidence for podocyte regeneration resulting from differentiation of progenitor cells within Bowman's capsule.


Subject(s)
Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Podocytes/cytology , Podocytes/metabolism , Animals , Bowman Capsule/cytology , Cell Differentiation/physiology , Humans , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Kidney Glomerulus/pathology , Podocytes/pathology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL